You are on page 1of 14

Malformations and Developmental Disorders - MC in the occiput

 (+) genetic and environmental influences (toxins, - Nasofrontal variants involving the orbit,
infections) ethmoid, or cribriform plate (“nasal
I. Neural Tube Defects glioma”) are also seen
o Midline malformations involving some  Spinal dysraphism or spina bifida
combination of neural tissue, meninges, and - MC NTD
overlying bone or soft tissue - Asymptomatic (occulta) or severe
o Collectively the most common CNS malformations (flattened disorganized segment of spinal
o Pathogenic Mechanisms cord associated with an overlying
 Failure of neural tube closure: secondary meningeal outpouching)
mesenchymal tissue defects stem from o Clinical Features
aberrant skeletal modeling around the  Frequency varies among ethnic groups (d/t
malformed tube polymorphisms in enzymes involved in folic
- Anencephaly acid metabolism)
- Myelomeningocele  Overall recurrence rate for NTD in subsequent
 Primary bone defects caused by abnormal pregnancies: 4-5%
axial mesoderm development: lead to  Folate deficiency during the first several
secondary CNS abnormalities weeks of gestation – risk factor
- Encephalocele - Folate supplementation can decrease
- Meningocele NTD risk but because neural tube closure
- Spina bifida is normally complete by day 28 of
o Morphology embryonic development (before most
 Anencephaly pregnancies are recognized), it must be
- Malformation of the anterior end of the given to women throughout their
neural tube reproductive years to be fully effective
- Absence of most of the brain and - May be due to effects on DNA
calvarium methylation
- Forebrain development disrupted at ~ 28 II. Forebrain Anomalies
days of gestation; remnant in its place is o Focal / diffuse
the area cerebrovasculosa (flattened o Abnormalities in the generation and migration of
disorganized brain tissue with admixed neurons
ependyma, choroid plexus, and o The pool of proliferating precursor cells in the
meningothelial cells) developing brain lies in the germinal matrix
- Posterior fossa structures may be spared adjacent to the ventricular system
depending on extent of skull deficit  Total number of neurons – determined by the
- Descending tracts associated with fraction of proliferating cells undergoing
disrupted structures are absent transition into migrating cells with each cell
 Myelomeningocele cycle
- Extension of CNS tissue through a o Paths of migration from the germinal matrix to
vertebral column defect the cerebral cortex:
- MC at lumbosacral region Migration Progenitor Cell Destiny
- (+) LE motor and sensory deficits, B&B Radial Excitatory neurons
dysfunction Tangential Inhibitory interneurons
- Complicated by superimposed infection o Pattern depends on gene mutation
of the cord d/t defective function of the o Changes either in complexity of brain surface,
thin overlying skin organization into lobes, cortical structure, or
 Encephalocele neuronal distribution
- Extrusion of malformed brain tissue  Brain volume changes: megalencephaly or
through a midline defect in the cranium microencephaly
chromosome X (mutant allele causes
Microencephaly male lethality; inn females, the process of
- More common X inactivation separates neurons into
- Typically with small HC those with a normal allele and those with
- Associated with chromosome the mutant allele)
abnormalities, fetal alcohol syndrome, - Doublecortin (DCX) – also in chromosome
and viral infection acquired in utero (HIV- X; mutation results in lissencephaly in
1, Zika virus) males and subcortical bandlike
- Reduction in the number of neurons that heterotopias in females (parallel layer of
reach the neocortex → simplification of gray matter – “double cortex”)
gyral folding  Holoprosencephaly
 Lissencephaly - Incomplete separation of the cerebral
- Reduction in the number of gyri hemispheres across the midline
- Extreme form: no gyri (agyria) - Severe forms with facial abnormalities
- Types (e.g. cyclopia); intrauterine Dx now
1  Smooth surface form possible
 Mutations disrupting cell - Less severe variants (arrhinencephaly) –
migration (e.g. in cytoskeletal absence of CN1 and related structures
motor proteins that drive - Associated with trisomy 13 or mutations
migration of neuroblasts) in genes encoding sonic hedgehog
2  Cobblestone surface form signaling pathway components
 Genetic alterations that  Agenesis of the corpus callosum
disrupt the stop signal for - Common
migration (depends on a set of
- Absence of interhemispheric white matter
glycosylated proteins) – MC
bundles
mutations in enzymes that
place sugars onto these - Radiology: misshapen lateral ventricles
proteins (bat wing deformity); on coronal whole
 Polymicrogyria mount sections of the brain, Probst
- Numerous small irregularly formed bundles of AP-oriented white matter can
cerebral convolutions with shallow sulci be demonstrated
- Cerebral cortex composed of 4 or fewer - Sometimes associated with intellectual
layers with fusion of the molecular layers disability but may also be found in normal
between gyri individuals
- Can be induced by localized tissue injury - Sporadic/familial
toward the end of neuronal migration - In isolation or associated with other
- Some are genetically determined malformations
(typically bilateral and symmetric) III. Posterior Fossa Anomalies
 Neuronal heterotopia o Primarily affect the brainstem and cerebellum +/-
- Group of migrational disorders defined by morphologic changes in other brain regions
collections of subcortical neurons in o Morphology
inappropriate locations along the  Arnold Chiari Malformation (Chiari type II)
pathway of migration - Small posterior fossa
- May be along the ventricular surface (no - Misshapen midline cerebellum with
migration at all) downward extension of the vermis
- Periventricular nodular heterotopias – through the foramen magnum
can be caused by mutations in the gene - Hydrocephalus
encoding filamin A (actin-binding protein - Lumbar myelomeningocele
responsible for assembly of complex - Other changes: medullary caudal
meshwork of filaments) located in displacement, tectal malformation,
aqueductal stenosis, cerebral Cerebrovascular Disease
heterotopia, hydromyelia  Hypoxia and Ischemia
 Chiari Type I Malformation o Brain – 1-2% BW but receives 15% of resting CO
- Less severe and accounts for 20% of O2 consumption
- Low lying cerebellar tonsils extend down o CBF remains ~ constant over a wide range of BP
into the vertebral canal and ICP d/t vascular resistance autoregulation
- Silent or symptomatic if with impaired o Ischemia → inappropriate release of excitatory AA
CSF flow and medullary compression NTs (e.g. glutamate) → damage neurons by
(correctable with neurosurgery) allowing excessive Ca influx through NMDA-type
 Dandy Walker Malformation glutamate receptors (excitotoxicity)
- Enlarged posterior fossa o Focal Cerebral Ischemia
- Cerebellar vermis absent or present in  Obstructive
rudimentary form anteriorly; replaced by  Collateral flow
a large midline cyst lined by ependyma - Circle of Willis
and contiguous with leptomeninges on its - Cortical-leptomeningeal anastomoses
outer surface (distal branches of ACA, MCA, and PCA)
- The cyst represents the expanded roofless - Little (if any) for the deep penetrating
4th ventricle in the absence of a normally- vessels
formed vermis  Embolism – MC affects the MCA (R = L)
- Commonly associated with dysplasia of - Cardiac mural thrombi
brainstem nuclei - Thromboemboli from arteries (MC
 Joubert Syndrome carotids)
- Hypoplasia of the cerebellar vermis with - Paradoxical embolism (CHD)
apparent elongation of the superior - Cardiac surgery
cerebellar peduncles and an altered - Tend to lodge where BVs branch / in
brainstem shape – molar tooth sign on areas of preexisting luminal stenosis
imaging - Shower embolization – fat embolization;
- Mutations in genes encoding components manifest as generalized cerebral
of the primary / nonmotile cilium dysfunction with disturbances of higher
IV. Syringomyelia and Hydromyelia cortical function and consciousness, often
o Hydromyelia - expansion of the ependyma-lined without localizing signs
central canal of the spinal cord - Widespread hemorrhagic lesions
o Syringomyelia / syrinx – formation of a fluid-filled involving white matter – characteristic of
cleft-like cavity inn the inner portion of the cord embolization of bone marrow after
 Syringobulbia – (+) brainstem extension trauma
 May be associated with a Chiari  Thrombotic occlusion
malformation, intraspinal tumors, or trauma - MC caused by acute change of vulnerable
 Histology atherosclerotic plaques (as in CAD)
- Destruction of the adjacent gray and - MC at carotid bifurcation, MCA origin,
white matter surrounded by dense and either ends of the basilar artery
reactive gliosis - Thrombi → progressive lumen narrowing,
 Manifests at 2nd/3rd decade of life anterograde extension, fragmentation,
 s/sx: isolated loss of pain and temperature and distal embolization
sensation in the UE  Inflammatory processes
- d/t disruption of the crossing anterior - Can lead to luminal narrowing
spinal commissural fibers - Infectious vasculitis of small and large
vessels occur with syphilis and TB,
immunosuppression and opportunistic
infection
- Non-infectious: PAN and other systemic - Loss of tinctorial characteristics of white
vasculitis, primary angiitis of the CNS and gray matter structures
 Other - Endothelial and glial cells (astrocytes
- Hypercoagulable states mainly) swell
- Dissecting aneurysm of extracranial - Myelinated fibers begin to disintegrate
arteries in the neck - Up to 48 h, neutrophilic emigration
- Drug abuse (amphetamine, heroin, progressively increases and then falls off
cocaine) 2. Subacute / evolving infarct
o +/- secondary hemorrhage - Phagocytic cells (circulating monocytes
 The brain has end-organ circulation with and activated microglia) evident at 48-72
limited collateral supply → occlusive brain h and become the predominant cell type
infarcts generally start as nonhemorrhagic in the ensuing 2-3 weeks
(pale/anemic) - Macrophages become stuffed with the
 Secondary hemorrhage can occur from products of myelin breakdown or blood
ischemia-reperfusion injury after spontaneous and may persist in the lesion for months
/ therapeutic dissolution / fragmentation of to years
the intravascular occlusive material - Reactive astrocytes and newly formed
- Hemorrhagic transformation vessels can be seen at the lesion
- Develops if the causative ischemic event periphery as early as 1 week after the
lasts long enough to damage small BVs in insult
the affected area - With liquefaction and phagocytosis,
- The resulting reperfusion hemorrhages astrocytes at the lesion edges
are largely petechial in nature, but may progressively enlarge, divide, and develop
be multiple or confluent a prominent network of cytoplasmic
o Morphology extensions
 Both gross and microscopic appearance 3. Healed infarct
change with time - After several months, the astrocytic
 Grossly, little change in appearance during response recedes, leaving behind a dense
the 1st 6 h of irreversible injury meshwork of glial fibers, new capillaries,
- By 48 h, the tissue becomes pale, soft, and perivascular CT
and swollen; gray-white junction - In the cerebral cortex, the cavity is
indistinct separated from the meninges and
- 2-10 d, the brain becomes gelatinous and subarachnoid space by a gliotic layer of
friable; the previously ill-defined border tissue (from molecular layer); pia and
between normal and infarcted tissue arachnoid unaffected
becomes more distinct as edema resolves - Stages progress from out to in
in the viable adjacent tissue  Hemorrhagic infarctions – additional blood
- 10d-3wk, tissue liquefies → fluid-filled extravasation and resorption
cavity that continues to expand until all of - In individuals on anticoagulation, may be
the dead tissue can be removed associated with intracerebral hematomas
 Microscopic - Venous infarcts – often hemorrhagic and
1. Acute infarct may occur after thrombotic occlusion of
- After the 1st 6-12 h, neurons in the the superior sagittal sinus or deep
affected area show eosinophilic neuronal cerebral veins; venous thrombosis risk
necrosis (increased eosinophilia of the increases with neoplasms, localized
cytoplasm followed by nuclear pyknosis infections, and hypercoagulable states
and karyorrhexis – dead red neurons) o Lacunar Infarcts (Lacunes)
- (+) cytotoxic and vasogenic edema  Hypertension affects the deep penetrating
arteries and arterioles supplying the BG,
hemispheric white matter, and BS; develop - Develop usually after severe hypotensive
arteriolosclerosis (400-900um) → thrombosis episodes; MC in patients resuscitated
and occlusion → lacunar infarcts after cardiac arrest
 Small cavitary infarcts; lake-like spaces (< Ꜫ
15mm wide)  Morphology
 Single / multiple - Global ischemia – the brain becomes
 Involve the putamen, GP, thalamus, internal edematous → widening of gyri, narrowing
capsule, deep white matter, caudate nucleus, of sulci
and pons - Poor gray-white demarcation
 Microscopically, (+) tissue loss surrounded by - Distinction from focal ischemic injury
gliosis based on overall pattern of brain
 Clinically silent or severe, depending on involvement (not nature of cellular
location pathology)
 Affected vessels may be associated with 1. Early changes (6-12 h)
widening of perivascular spaces without - Dead red neurons
tissue infarction (etat crible) - Acute changes later in astrocytes and
o Global Cerebral Ischemia / Hypoxia oligodendrocytes
 Generalized reduction in cerebral perfusion / 2. Subacute changes (24 h – 2 weeks)
blood oxygen - Tissue necrosis, influx of macrophages,
 Reversible if mild (transient post-ischemic vascular proliferation, and reactive gliosis
confusional state); irreversible in some cases 3. Repair (after 2 weeks)
(diffuse hypoxic/ischemic encephalopathy) - Removal of necrotic tissue, loss of normal
 Sensitivity to hypoxia: neurons > glial cells CNS architecture, and gliosis
- Oligodendrocytes and astrocytes are also - Laminar necrosis – pattern produced in
vulnerable the cerebral neocortex; neuronal loss and
- Most sensitive neurons: pyramidal gliosis uneven (some layers preserved,
neurons in the hippocampus (area CA1 – others destroyed)
Sommer sector), cerebellar Purkinje cells,  Intracranial Hemorrhage
pyramidal neurons in the cerebral cortex Hemorrhage MC Associated With
(layers III and IV) Epidural Trauma
 Widespread neuronal death; surviving Subdural
patients often in persistent vegetative state Intraparenchymal Underlying CVD
 Others experience brain death Subarachnoid
- Irreversible diffuse cortical injury o Intraparenchymal Hemorrhage
(isoelectric / flat EEG)  Spontaneous/non-traumatic – MC in mid-late
- BS damage adult life
- Absent cerebral perfusion  Types and their major causes
- If maintained on mech vent, the brain - Ganglionic (BG, thalamus) – hypertension
gradually undergoes widespread - lobar (cerebral hemispheres) – cerebral
liquefaction (“respirator brain”) amyloid angiopathy
 Border zone / Watershed infarcts  Contributory factors: systemic coagulation
- At areas in the most distal reaches of the disorders, neoplasms, vasculitis, aneurysms,
arterial blood supply and vascular malformations
- MC at MCA-ACA – a few cm lateral to the  Hypertension - MC associated with deep
interhemispheric fissure; damage results hemorrhages (> 50% clinically significant, 15%
to a cortical wedge-shaped infarct with mortality)
secondary hemorrhagic transformation; - MC in the putamen; others include
often bilateral thalamus, pons, cerebellar hemispheres
- Arteriolar walls affected by hyaline - Other causes: rupture of a primary
change are thickened but more prone to intracerebral hemorrhage into the
rupture (most prominent in the BG and ventricular system, vascular
subcortical white matter) malformation, hematologic disturbances,
and tumors
 Cerebral amyloid angiopathy (CAA)  However, generally MC cause is trauma
- Amyloidogenic peptides deposited in the  Saccular aneurysm – MC type of intracranial
walls of medium and small caliber aneurysm
meningeal, cortical, and cerebellar vessels - Other aneurysm types: atherosclerotic
- Involved vessels are rigid (fusiform; MC at basilar artery), mycotic,
- Hyaline consists of beta amyloid rather traumatic, and dissecting
than collagen - 90% near the major arterial branch points
- Amyloid deposition weakens the vessel in the anterior circulation
wall → hemorrhage – (+) microbleeds  Pathogenesis: Saccular aneurysms
- Increased risk of bleed with the presence - Absence of smooth muscle and intimal
of an Ꜫ2 or Ꜫ4 allele elastic lamina – suggestive of being
- AD forms associated with APP mutations developmental anomalies
– encodes precursor for A-beta peptides - MC occur sporadically, but genetics are
 Cerebral AD arteriopathy with subcortical considered; > risk in:
infarcts and leukoencephalopathy (CADASIL) a. 1st degree relatives
- Mutations in the NOTCH3 gene b. Mendelian disorders – AD polycystic
- Recurrent small vessel infarct and kidney disease, Ehlers-Danlos
dementia syndrome type IV, NF1, Marfan
- Initially white matter changes Syndrome)
- MC presents at 35 y/o c. Fibromuscular dysplasia of
 Morphology extracranial arteries
- Acute primary intraparenchymal d. Coarctation of aorta
hemorrhages – (+) central core of clotted - Other predisposing factors: cigarette
blood that compresses the adjacent smoking, hypertension
parenchyma → secondary infarction - Not present at birth (even if sometimes
(anoxic changes and edema) referred to as congenital); develop over
- Edema resolves, hemosiderin and lipid time d/t an underlying media defect
laden macrophages appear, and  Morphology
proliferation of reactive astrocytes is seen - Unruptured saccular aneurysm – thin-
at the lesion periphery walled outpouching usually at an arterial
- Old hemorrhages show areas of branch point along the circle of Willis or a
parenchymal cavitary destruction with a major vessel just beyond
rim of brownish discoloration - Few mm to 2-3 cm; bright red shiny
 Clinical Features surface and a thin translucent wall
- Devastating if large involvement or with - Atheromatous plaques / calcifications /
extension into the ventricular system thrombi may be found in the wall / lumen
- If smaller regions are affected, may be of the aneurysm
clinically silent or evolve like an infarct; - Evidence of prior hemorrhage: brownish
over weeks/months, hematoma is discoloration of the adjacent brain and
gradually removed, +/- clinical meninges
improvement - Neck of aneurysm wide or narrow
o Subarachnoid Hemorrhage - Rupture usually occurs at the apex of the
 Spontaneous MC d/t rupture of a saccular / sac → extravasation of blood in the
berry aneurysm in a cerebral artery subarachnoid space / parenchyma
- Arterial wall adjacent to the aneurysm - Vessels involved: subarachnoid space /
neck – (+) intimal thickening and media brain parenchyma
attenuation - Composed of greatly enlarged BVs
- Smooth muscle and intimal elastic lamina separated by gliotic tissue, often with
do not extend into the neck and are evidence of prior hemorrhage
absent from the aneurysm sac itself - Some are arteries with
(made up of thickened hyalinized intima duplication/fragmentation of the internal
and a covering of adventitia) elastic lamina, while others show marked
 Clinical Features: rupture thickening or partial replacement of the
- MC in the 5th decade, F > M media by hyalinized CT
- Occur anytime but 1/3 cases associated 2. Cavernous malformations
with acute increases in ICP, such as with - Distended loosely organized vascular
straining at stool or sexual orgasm channels arranged back-to-back with
- Blood under arterial pressure is forced collagenized walls of variable thickness
into the subarachnoid space → sudden - (-) brain parenchyma in between vessels
excruciating HA - MC in the cerebellum, pons, and
- 25-50% die with the first rupture; those subcortical regions
who survive often improve and recover - Low flow (no shunting)
consciousness in minutes - Foci of old hemorrhage, infarction, and
- Repeat bleed common in survivors and calcification frequently surround the
unpredictable in timing abnormal vessels
- Clinical consequences of blood in the  Clinical Features: AVM
subarachnoid space: - M>F
a. Acute (hours – days) – increased risk - Often presents at 10-30 y/o as a seizure
of additional ischemic injury from d/o, an ICH, or a SAH
vasospasm affecting vessels bathed in - MC at posterior branches of MCA
extravasated blood; most significant - Large AVM in the newborn can lead to
in basal SAH (major vessels of the CHF d/t shunting, especially if the vein of
circle of Willis can be involved); Galen is involved
mediators include endothelin, NO, **cavernous malformations – familial forms
arachidonic acid metabolites common (hallmark: multiple lesions;
b. Late – associated with healing penetrant AD)
processes; meningeal fibrosis and  Vascular Dementia
scarring – may obstruct CSF flow o Individuals who suffer multiple bilateral gray
o Vascular Malformations (cortex, thalamus, BG) and white (centrum
 Types: AVM, cavernous malformations, semiovale) matter infarcts over months-years
capillary telangiectasias, venous angiomas may develop a distinct clinical syndrome of:
- 1st 2 associated with hemorrhage risk  Dementia
 AVM frequently associated with activating  Gait abnormalities
somatic mutations in the KRAS oncogene  Pseudobulbar signs with FND
within the endothelial cells that line the o d/t multifocal vascular disease
malformed vessels (dysregulated RAS  Cerebral atherosclerosis
signaling)  Vessel thrombosis / embolization from
 Morphology carotid vessels or the heart
1. AVM  Cerebral arteriolosclerosis from chronic
- Tangled networks of worm-like vascular hypertension
channels with high BF d/t prominent o Binswanger disease – injury pattern preferentially
pulsatile AV shunting involves large areas of the subcortical white
matter with myelin and axon loss (subcortical  In immunosuppressed individuals, purulent
white matter dementia) meningitis may be caused by other infectious
organisms such as Klebsiella or anaerobes –
atypical clinical course and CSF findings
 Morphology
- Exudate evident within the
leptomeninges over the brain surface
- Meningeal vessels engorged and
CNS INFECTIONS prominent
 Infections may damage the nervous system directly - H. influenza meningitis – basal;
by the infectious agent or indirectly by microbial Pneumococcal meningitis – densest over
toxin, inflammatory responses, and immune- cerebral convexities near the sagittal
mediated mechanisms sinus
 Routes of microbe entry - From areas of greatest accumulation,
o Hematogenous – MC; often via arterial tracts of pus follow along BVs on the brain
circulation; venous spread occurs retrograde via surface
anastomoses with facial veins - If fulminant, +/- ventriculitis; lack of BBB
o Direct inoculation – traumatic or may be in the choroid plexus may also be the
associated with congenital malformations such as portal for CSF involvement of blood-borne
meningomyelocele infections
o Local extension – air sinuses, teeth, skull, - Neutrophils fill the subarachnoid space in
vertebrae severely affected areas; predominant
o PNS – viral spread like with rabies and herpes around leptomeningeal BVs in < severe
zoster cases
I. Acute Meningitis - In fulminant meningitis, the inflammatory
 Acute pyogenic – bacterial cells infiltrate the walls of the
 Aseptic – acute/subacute viral leptomeningeal veins and may extend
 Chronic – tuberculous, spirochetal, into the brain substance (cerebritis)
cryptococcal - Secondary vasculitis and venous
1. Acute Pyogenic (Bacterial) Meningitis thrombosis may lead to hemorrhagic
 E. coli and GBS – neonates cerebral infarction
 S. pneumoniae and L. monocytogenes – - Leptomeningeal fibrosis may follow
elderly pyogenic meningitis and cause
 N. meningitidis – adolescents and young hydrocephalus; in pneumococcal
adults meningitis, large quantities of the
 Systemic signs of infection superimposed on capsular polysaccharide of the organism
symptoms related to meningeal irritation and produce a gelatinous exudate that
neurologic impairment promotes arachnoid fibrosis (chronic
 CSF: 90,000 neutrophils per mm 3, increased adhesive arachnoiditis)
CSF pressure, increased protein, reduced 2. Acute Aseptic (Viral) Meningitis
glucose  (-) bacterial culture but with s/sx of meningitis
 Fatal if untreated  Generally viral but may be bacterial /
 Complication: Waterhouse-Frederichsen rickettsial / autoimmune
Syndrome  Less fulminant clinical course
- Results from meningitis associated  CSF: lymphocytic pleocytosis, moderate
septicemia and hemorrhagic infarction of protein increase, glucose ~ normal
the adrenal glands  Self-limited; symptomatic tx; etiology rarely
- MC with meningococcal and identified – MC enteroviruses (80%)
pneumococcal meningitis
 Clinical picture may develop after rupture of  Bacterial (and rarely fungal) infections of the
an epidermoid cyst into the subarachnoid skull bones or air sinuses can spread to the
space or with chemical meningitis subdural space
- CSF: sterile, pleocytosis with neutrophils,  +/- mass effect and/or thrombophlebitis of
increased protein, normal glucose the bridging veins – can cause occlusion and
II. Acute Focal Suppurative Infections infarction of the brain
o Pyogenic bacteria / fungi  FNDs, febrile, HA, neck stiffness
1. Brain Abscess  CSF profile similar to that of brain abscess
 Localized focus of necrosis of brain tissue with  Full recovery with prompt Dx and Tx; only
accompanying inflammation, MC because of residuum is a thickened dura
bacterial infection 3. Extradural Abscess
 Causes  Commonly associated with osteomyelitis
- Direct implantation  MC from an adjacent focus of infection
- Local extension (mastoiditis, paranasal (sinusitis) or post-op
sinusitis)  If at the spinal epidural space, it may cause
- Hematogenous spread (heart, lungs, SCC – neurosurgical emergency
bone; bacteremia from dental III. Chronic Bacterial Meningoencephalitis
procedures) o Causes: Mycobacterium tuberculosis, Treponema
 Predisposing conditions pallidum, and Borrelia
- Acute bacterial endocarditis 1. Tuberculosis
- CHD with R to L shunting and loss of  May be part of active disease elsewhere in
pulmonary filtration the body or appear in isolation after seeding
- Chronic pulmonary sepsis (bronchiectasis) from silent lesions (MC from lungs)
- Systemic disease with  May involve the meninges or the brain itself
immunosuppression  Morphology
 MC organisms in non-immunosuppressed a. Diffuse meningoencephalitis
patients: streptococci and staphylococci - MC pattern
 Morphology - Subarachnoid space contains a
- Discrete lesions with central liquefactive gelatinous / fibrinous exudate
necrosis surrounded by brain swelling characteristically involving the base of the
- (+) exuberant granulation tissue with brain, effacing the cisterns and encasing
neovascularization – newly formed cranial nerves
vessels are abnormally permeable → - +/- discrete white areas of inflammation
marked vasogenic edema scattered over the leptomeninges
- In well-established lesions, a collagenous - Involved areas contain a mixed
capsule is produced by fibroblasts from inflammatory infiltrate with lymphocytes,
BV walls; outside the fibrous capsule is a plasma cells, and macrophages
zone of reactive gliosis with numerous - Florid cases show well-formed
gemistocytic astrocytes granulomas with caseous necrosis and
 Clinical Features giant cells
- Progressive FNDs; s/sx related to - Arteries running through the
increased ICP subarachnoid space may show
- CSF: high WBC count, increased protein obliterative endarteritis and marked
concentration, normal glucose intimal thickening
- Complications: herniation, rupture with - May spread to the choroid plexus and
ventriculitis / meningitis, venous sinus ependymal surface, travelling through the
thrombosis CSF
- Low mortality with proper tx
2. Subdural Empyema
- In long-standing cases, a dense fibrous - Insidious but progressive cognitive
adhesive arachnoiditis may develop (most impairment associated with mood
conspicuous around the brain base) alterations (including delusions of
- +/- hydrocephalus grandeur) that terminate in severe
b. Tuberculoma dementia (general paresis of the insane)
- Well-circumscribed intraparenchymal - Parenchymal damage of the cerebral
mass, +/- meningitis cortex – common in the frontal lobe
- If several cm in diameter, may cause - Loss of neurons, proliferation of
significant mass effect microglia, gliosis, and iron deposits
- (+) central caseous necrosis (Prussian blue stain)
- Calcified if inactive - Spirochetes in tissue sections
 Clinical Features 3. Tabes dorsalis
- HA, malaise, mental confusion, vomiting - Damage to sensory axons in the dorsal
- CSF: pleocytosis made up of mononuclear roots
cells +/I neutrophils, increased protein, - Impaired joint position sense, locomotor
moderately decreased / normal glucose ataxia
- Most serious complications of chronic - Loss of pain sensation → Charcot joint
tuberculous meningitis: - Lightning pain, areflexia
a. Arachnoid fibrosis → hydrocephalus - Loss of both axons and myelin in the
b. Obliterative endarteritis → arterial dorsal roots, with pallor and atrophy in
occlusion and brain infarction the dorsal columns of the SC
- If the spinal cord subarachnoid space is - Organisms not demonstratable
involved, nerve roots may be affected 3. Neuroborreliosis (Lyme Disease)
- Tuberculomas – d/dx: CNS tumors  Caused by the spirochete Borrelia burgdorferi
- < host reaction in AIDS patients (transmitted by Ixodes ticks)
2. Neurosyphilis  Neurologic symptoms highly variable: aseptic
 Manifestation of the tertiary stage of syphilis meningitis, facial nerve palsies,
 Occurs in 10% individuals with untreated polyneuropathies, encephalopathy
infection  Focal proliferation of microglial cells in the
 Patterns: meningovascular / paretic / tabes brain; scatted extracellular organisms
dorsalis IV. Viral Meningoencephalitis
 d/t impaired cell-mediated immunity, > risk in o Some viruses have a propensity to infect the
patients with HIV (especially acute syphilitic nervous system (cell type or brain area)
meningitis or meningovascular disease) o Consider latency
 Morphology o Systemic viral infections in the absence of direct
1. Meningovascular neurosyphilis evidence of viral penetration into the CNS may be
- Chronic meningitis involving the base of followed by an immune-mediated disease (e.g.
the brain, the cerebral convexities, and perivenous demyelination)
spinal leptomeninges 1) Arthropod-borne
- +/- obliterative endarteritis (Heubner  Arboviruses – important cause of epidemic
arteritis) with distinctive perivascular encephalitis; animal hosts, insect vectors
inflammatory reaction rich in plasma cells  Neurologic deficits: generalized (seizures,
and lymphocytes confusion, delirium, stupor/coma) and focal
- Cerebral gummas (plasma cell – rich mass  Involvement of the spinal cord in West Nile
lesions) may also occur in the meninges encephalitis can lead to a polio-like syndrome
and extend into the parenchyma with paralysis
2. Paretic neurosyphilis  CSF: colorless, slightly increased pressure,
- Invasion of the brain by T. pallidum increased protein, normal glucose;
neutrophilic pleocytosis → lymphocytosis
 Morphology  In utero infection → periventricular necrosis
- Perivascular accumulation of lymphocytes → severe brain destruction → microcephaly
+/- neutrophils and periventricular calcification
- Multiple foci of necrosis of gray and white  Morphology (immunosuppressed)
matter - Subacute encephalitis +/- CMV inclusion-
- Single cell neuronal necrosis with bearing cells
phagocytosis of the debris - Localize in the subependymal regions →
(neuronophagia) severe hemorrhagic necrotizing
- Microglial cells form small aggregates: ventriculoencephalitis and choroid plexitis
microglial nodules - Can also attack the lower SC and roots →
- Severe cases: necrotizing vasculitis with radiculoneuritis
focal hemorrhages - May affect any CNS cell
2) Herpes Simplex Virus Type 1 - Infection confirmed with
 Encephalitis MC in children and young adults; immunohistochemistry
10% have a history of prior herpetic infection - Conventional light microscopy: prominent
 Typical presenting sx: changes in mood, enlarged cells with intranuclear and
memory, and behavior intracytoplasmic inclusions
 Effectively treated with antivirals 6) Poliomyelitis
 Morphology  In non-immunized individuals, poliovirus
- Starts in and most severe at the inferior infection causes subclinical / mild
and medial temporal lobes and the orbital gastroenteritis but rarely can secondarily
gyri of the frontal lobes invade the nervous system
- Necrotizing and hemorrhagic  Morphology
- Perivascular inflammatory infiltrates - Acute cases show mononuclear cell
- Cowdry type A intranuclear viral perivascular cuffs and neuronophagia of
inclusions may be found in neurons and the AHCs (may extend posteriorly, +/-
glia cavitation)
- If slowly evolving, brain involvement is  Clinical Features
more diffuse - Initial: meningeal irritation and CSF
3) HSV Type 2 picture consistent with aseptic meningitis
 Adults: meningitis → +/- progression to involve the spinal
 Neonates: encephalitis – vaginal delivery in cord → flaccid paralysis (may involve the
women with active primary HSV genital respiratory muscles)
infections - Myocarditis can sometimes complicate
 HIV patients: acute hemorrhagic and the acute infection
necrotizing encephalitis - Postpolio syndrome can develop in
4) Varicella Zoster Virus patients 25-35 years after resolution of
 Primary infection (chickenpox) – childhood the initial illness; progressive weakness
exanthems without neurologic involvement with decreased muscle mass and pain;
 Reactivation in adults: shingles / herpes superimposed loss of remaining motor
zoster neurons without evidence of viral re-
- Self-limited but there may be activation
postherpetic neuralgia syndrome (after 60 7) Rabies
y/o) – vaccine preventable  Severe encephalitis
5) Cytomegalovirus  From a bite of a rabid animal; exposure to
 In fetuses and immunosuppressed individuals certain bat species without a bite can also
(common opportunistic infection in AIDS) cause it
 Morphology
- External brain: intense edema and - Among CNS cells, only microglia express
vascular congestion both CD4 and CCR5/CXCR4 receptors
- Widespread neuronal degeneration and - Chronic: HIV encephalitis
inflammation most severe in the  Immune reconstitution Inflammatory
brainstem Syndrome (IRIS)
- BG, DC and DRG may be involved - Identified in some patients after effective
- Negri bodies – pathognomonic; tx; paradoxical deterioration after starting
cytoplasmic round/oval eosinophilic therapy
inclusions in pyramidal neurons of the - Exuberant reconstituted inflammatory
hippocampus and Purkinje cells of the response; intense inflammation with an
cerebellum (sites of no inflammation); influx of CD8+ lymphocytes
viruses can be detected within by  Morphology
ultrastructural and immunohistochemical - HIV encephalitis: chronic inflammatory
methods reaction associated with widely
 Clinical Features distributed microglial nodules (contain
- Virus enters the CNS by ascending along macrophage-derived multinucleated giant
peripheral nerves from the wound site cells)
- Incubation period: 1-3 months - +/- foci of tissue necrosis and reactive
(depending on distance between wound gliosis
and brain) - Some of the microglial nodules are found
- Disease begins with nonspecific near small BVs – abnormally prominent
symptoms: malaise, HA, fever; if with endothelial cells and perivascular foamy /
local paresthesia around the wound – pigment-laden macrophages
diagnostic - Changes prominent in the subcortical
- As the infection advances, the affected white matter, diencephalon, and
individual exhibits extraordinary CNS brainstem
excitability (slight touch is painful and - HIV can be detected CD4+ microglia and
produces violent motor responses or mononuclear/multinucleated
convulsions) macrophages
- Contracture of pharyngeal muscles on  Clinical Features
swallowing → mouth foaming; aversion to - Cognitive changes (mild or HIV-associated
swallowing and water (hydrophobia) dementia) – HIV-associated
- Meningeal irritation, flaccid paralysis neurocognitive disorders (HAND) – most
- Alternating periods of mania and stupor closely related to inflammatory activation
progress to coma and eventual death of microglial and perivascular
from respiratory failure macrophages
8) HIV - Mechanisms of neuronal dysfunction:
 Direct effects of the virus on the nervous cytokine action, HIV-derived protein
system, opportunistic infections, and primary toxicity, anti-HIV tx neurotoxicity,
CNS lymphoma (EBV-positive B-cell tumor) – accelerated aging, aberrant synaptic
decreased frequency d/t current effective pruning
antiretroviral tx 9) Progressive Multifocal Leukoencephalopathy
 HIV aseptic meningitis – occurs in 1-2weeks of  Encephalitis caused by the JC polyomavirus –
seroconversion in 10% patients preferentially infects oligodendrocytes –
- (+) HIV specific antibodies; virus can be demyelination is the principal pathologic
isolated from CSF effect
- Early phase: mild lymphocytic meningitis,  Often exclusive in immunosuppressed
perivascular inflammation, myelin loss individuals (chronic lymphoproliferative /
myeloproliferative illnesses,
immunosuppressive chemotherapy,  Cryptococcal meningitis
granulomatous diseases, AIDS) - Common in AIDS
 Most people have serologic evidence of - May be fulminant and fatal in 2 weeks or
exposure to JC virus by 14 y/o, but primary indolent (months/years)
infection asymptomatic - CSF may contain few cells but usually has
 PML results from viral reactivation with a high protein concentration
immunosuppression - Mucoid encapsulated yeasts can be
 Clinically: affected individuals develop focal visualized in the CSF with special stains or
and relentlessly progressive neurologic s/sx detected indirectly using assays for
 Imaging: extensive multifocal cerebral / cryptococcal antigens
cerebellar white matter lesions - MC C. neoformans; other: C. gattii –
 Morphology formation of cryptococcomas
- Patches of irregular ill-defined white VI. Other Infections of the Nervous System
matter injury ranging from mm to large o Protozoal diseases (malaria, toxoplasmosis,
confluent lesions amebiasis, trypanosomiasis), rickettsial infections
- Area of demyelination MC in a subcortical (typhus, Rocky Mountain spotted fever), and
region; at the center are sheets of lipid- metazoal diseases (cysticercosis, echinococcosis)
laden macrophages and a reduced 1) Cerebral toxoplasmosis
number of axons  Common opportunistic infection in HIV
- Greatly enlarged oligodendrocyte nuclei  Subacute, evolves during a 1–2-week period
with glassy amphophilic viral inclusions –  Focal/diffuse
immunohistochemistry; seen at lesion  CT / MR: multiple ring-enhancing lesions
edge - Non-specific; also in CNS lymphoma, TB,
- Bizarre giant astrocytes with >1 irregular and fungal infections
hyperchromatic nuclei are intermixed  In non-immunosuppressed, the impact of
with more typical reactive astrocytes toxoplasmosis is most often seen when
- Infection of granule cell neurons in the primary maternal infection occurs early in
cerebellum – rare pregnancy
V. Fungal Meningoencephalitis - Spread to the brain of the developing
o MC in immunocompromised individuals; fetus and cause severe damage in the
widespread hematogenous dissemination reaches form of multifocal necrotizing lesions that
the brain may calcify
o MC: Candida, Mucor, Aspergillus, Cryptococcus  Treatable with antibiotics with early dx
o Direct extension may also occur, especially in  (+) brain abscess – most often in the cerebral
mucormycosis with DM cortex and deep gray nuclei
o Forms of CNS fungal infection: chronic meningitis, - Acute lesions: central necrosis, petechial
vasculitis, and parenchymal invasion hemorrhages surrounded by acute and
 Vasculitis – Mucormycosis and Aspergillosis chronic inflammation, macrophage
MC; vascular thrombosis can produce infiltration, and vascular proliferation
hemorrhagic infarction - Free tachyzoites and encysted bradyzoites
 Parenchymal invasion – MC as granulomas or may be found at the periphery of the
abscesses; Candida and Cryptococcus MC necrotic foci
- Candidiasis: multiple microabscesses +/- - Seen on routine H&E or Giemsa stains,
granuloma formation but more recognized with
- Cryptococcus: parenchymal aggregates immunohistochemical methods
typically found within expanded - BVs near the lesions: intimal
perivascular (Virchow-Robin) spaces and proliferation / frank vasculitis with
are associated with minimal/no fibrinoid necrosis or thrombosis
inflammation or gliosis 2) Cerebral amebiasis
 Rapidly fatal necrotizing encephalitis from
Naegleria species infection
 Chronic granulomatous meningoencephalitis
associated with Acanthamoeba
 Amebae – difficult to distinguish
morphologically from activated macrophages
 Methenamine silver or PAS can help in
visualization
3) Cerebral malaria
 Complication of infection by Plasmodium
falciparum
 Result of sticking of infected RBCs to the
inflamed vascular endothelium
 Reduced cerebral BF a
 Ataxia, seizures, and coma in the acute phase;
long term cognitive deficits in 20% children

You might also like